Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
medRxiv ; 2023 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-37732212

RESUMO

SARS-CoV-2 is spread through exhaled breath of infected individuals. A fundamental question in understanding transmission of SARS-CoV-2 is how much virus an individual is exhaling into the environment while they breathe, over the course of their infection. Research on viral load dynamics during COVID-19 infection has focused on internal swab specimens, which provide a measure of viral loads inside the respiratory tract, but not on breath. Therefore, the dynamics of viral shedding on exhaled breath over the course of infection are poorly understood. Here, we collected exhaled breath specimens from COVID-19 patients and used RTq-PCR to show that numbers of exhaled SARS-CoV-2 RNA copies during COVID-19 infection do not decrease significantly until day 8 from symptom-onset. COVID-19-positive participants exhaled an average of 80 SARS-CoV-2 viral RNA copies per minute during the first 8 days of infection, with significant variability both between and within individuals, including spikes over 800 copies a minute in some patients. After day 8, there was a steep drop to levels nearing the limit of detection, persisting for up to 20 days. We further found that levels of exhaled viral RNA increased with self-rated symptom-severity, though individual variation was high. Levels of exhaled viral RNA did not differ across age, sex, time of day, vaccination status or viral variant. Our data provide a fine-grained, direct measure of the number of SARS-CoV-2 viral copies exhaled per minute during natural breathing-including 312 breath specimens collected multiple times daily over the course of infection-in order to fill an important gap in our understanding of the time course of exhaled viral loads in COVID-19.

2.
J Neurosci ; 42(34): 6506-6517, 2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-35906072

RESUMO

Schwann cells play a critical role after peripheral nerve injury by clearing myelin debris, forming axon-guiding bands of Büngner, and remyelinating regenerating axons. Schwann cells undergo epigenomic remodeling to differentiate into a repair state that expresses unique genes, some of which are not expressed at other stages of Schwann cell development. We previously identified a set of enhancers that are activated in Schwann cells after nerve injury, and we determined whether these enhancers are preprogrammed into the Schwann cell epigenome as poised enhancers before injury. Poised enhancers share many attributes of active enhancers, such as open chromatin, but are marked by repressive histone H3 lysine 27 (H3K27) trimethylation rather than H3K27 acetylation. We find that most injury-induced enhancers are not marked as poised enhancers before injury indicating that injury-induced enhancers are not preprogrammed in the Schwann cell epigenome. Injury-induced enhancers are enriched with AP-1 binding motifs, and the c-JUN subunit of AP-1 had been shown to be critical to drive the transcriptional response of Schwann cells after injury. Using in vivo chromatin immunoprecipitation sequencing analysis in rat, we find that c-JUN binds to a subset of injury-induced enhancers. To test the role of specific injury-induced enhancers, we focused on c-JUN-binding enhancers upstream of the Sonic hedgehog (Shh) gene, which is only upregulated in repair Schwann cells compared with other stages of Schwann cell development. We used targeted deletions in male/female mice to show that the enhancers are required for robust induction of the Shh gene after injury.SIGNIFICANCE STATEMENT The proregenerative actions of Schwann cells after nerve injury depends on profound reprogramming of the epigenome. The repair state is directed by injury-induced transcription factors, like JUN, which is uniquely required after nerve injury. In this study, we test whether the injury program is preprogrammed into the epigenome as poised enhancers and define which enhancers bind JUN. Finally, we test the roles of these enhancers by performing clustered regularly interspaced short palindromic repeat (CRISPR)-mediated deletion of JUN-bound injury enhancers in the Sonic hedgehog gene. Although many long-range enhancers drive expression of Sonic hedgehog at different developmental stages of specific tissues, these studies identify an entirely new set of enhancers that are required for Sonic hedgehog induction in Schwann cells after injury.


Assuntos
Proteínas Hedgehog , Traumatismos dos Nervos Periféricos , Proteínas Proto-Oncogênicas c-jun , Animais , Feminino , Proteínas Hedgehog/metabolismo , Masculino , Camundongos , Bainha de Mielina/metabolismo , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ratos , Células de Schwann/metabolismo , Fator de Transcrição AP-1/metabolismo
4.
Front Immunol ; 11: 1185, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32612606

RESUMO

Indoleamine 2, 3-dioxygenase 1 (IDO; IDO1; INDO) is a rate-limiting enzyme that metabolizes the essential amino acid, tryptophan, into downstream kynurenines. Canonically, the metabolic depletion of tryptophan and/or the accumulation of kynurenine is the mechanism that defines how immunosuppressive IDO inhibits immune cell effector functions and/or facilitates T cell death. Non-canonically, IDO also suppresses immunity through non-enzymic effects. Since IDO targeting compounds predominantly aim to inhibit metabolic activity as evidenced across the numerous clinical trials currently evaluating safety/efficacy in patients with cancer, in addition to the recent disappointment of IDO enzyme inhibitor therapy during the phase III ECHO-301 trial, the issue of IDO non-enzyme effects have come to the forefront of mechanistic and therapeutic consideration(s). Here, we review enzyme-dependent and -independent IDO-mediated immunosuppression as it primarily relates to glioblastoma (GBM); the most common and aggressive primary brain tumor in adults. Our group's recent discovery that IDO levels increase in the brain parenchyma during advanced age and regardless of whether GBM is present, highlights an immunosuppressive synergy between aging-increased IDO activity in cells of the central nervous system that reside outside of the brain tumor but collaborate with GBM cell IDO activity inside of the tumor. Because of their potential value for the in vivo study of IDO, we also review current transgenic animal modeling systems while highlighting three new constructs recently created by our group. This work converges on the central premise that maximal immunotherapeutic efficacy in subjects with advanced cancer requires both IDO enzyme- and non-enzyme-neutralization, which is not adequately addressed by available IDO-targeting pharmacologic approaches at this time.


Assuntos
Neoplasias Encefálicas/imunologia , Encéfalo/enzimologia , Glioblastoma/imunologia , Tolerância Imunológica/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Animais , Neoplasias Encefálicas/enzimologia , Modelos Animais de Doenças , Glioblastoma/enzimologia , Humanos , Neoplasias/enzimologia , Neoplasias/imunologia
5.
Genes Dev ; 34(15-16): 1089-1105, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32616519

RESUMO

The circadian clock is encoded by a negative transcriptional feedback loop that coordinates physiology and behavior through molecular programs that remain incompletely understood. Here, we reveal rhythmic genome-wide alternative splicing (AS) of pre-mRNAs encoding regulators of peptidergic secretion within pancreatic ß cells that are perturbed in Clock-/- and Bmal1-/- ß-cell lines. We show that the RNA-binding protein THRAP3 (thyroid hormone receptor-associated protein 3) regulates circadian clock-dependent AS by binding to exons at coding sequences flanking exons that are more frequently skipped in clock mutant ß cells, including transcripts encoding Cask (calcium/calmodulin-dependent serine protein kinase) and Madd (MAP kinase-activating death domain). Depletion of THRAP3 restores expression of the long isoforms of Cask and Madd, and mimicking exon skipping in these transcripts through antisense oligonucleotide delivery in wild-type islets reduces glucose-stimulated insulin secretion. Finally, we identify shared networks of alternatively spliced exocytic genes from islets of rodent models of diet-induced obesity that significantly overlap with clock mutants. Our results establish a role for pre-mRNA alternative splicing in ß-cell function across the sleep/wake cycle.


Assuntos
Processamento Alternativo , Relógios Circadianos/genética , Exocitose , Glucose/metabolismo , Secreção de Insulina/genética , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/fisiologia , Animais , Proteínas CLOCK/genética , Proteínas CLOCK/fisiologia , Células Cultivadas , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Guanilato Quinases/genética , Guanilato Quinases/metabolismo , Homeostase , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Proteínas Nucleares/fisiologia , Obesidade/genética , Obesidade/metabolismo , Proteína 25 Associada a Sinaptossoma/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Fatores de Transcrição/fisiologia
6.
Dis Model Mech ; 13(2)2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31582396

RESUMO

Limb-girdle muscular dystrophy type 2C is caused by autosomal recessive mutations in the γ-sarcoglycan (SGCG) gene. The most common SGCG mutation is a single nucleotide deletion from a stretch of five thymine residues in SGCG exon 6 (521ΔT). This founder mutation disrupts the transcript reading frame, abolishing protein expression. An antisense oligonucleotide exon-skipping method to reframe the human 521ΔT transcript requires skipping four exons to generate a functional, internally truncated protein. In vivo evaluation of this multi-exon skipping, antisense-mediated therapy requires a genetically appropriate mouse model. The human and mouse γ-sarcoglycan genes are highly homologous in sequence and gene structure, including the exon 6 region harboring the founder mutation. Herein, we describe a new mouse model of this form of limb-girdle muscular dystrophy generated using CRISPR/Cas9-mediated gene editing to introduce a single thymine deletion in murine exon 6, recreating the 521ΔT point mutation in Sgcg These mice express the 521ΔT transcript, lack γ-sarcoglycan protein and exhibit a severe dystrophic phenotype. Phenotypic characterization demonstrated reduced muscle mass, increased sarcolemmal leak and fragility, and decreased muscle function, consistent with the human pathological findings. Furthermore, we showed that intramuscular administration of a murine-specific multiple exon-directed antisense oligonucleotide cocktail effectively corrected the 521ΔT reading frame. These data demonstrate a molecularly and pathologically suitable model for in vivo testing of a multi-exon skipping strategy to advance preclinical development of this genetic correction approach.


Assuntos
Éxons/genética , Edição de Genes , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Animais , Sequência de Bases , Modelos Animais de Doenças , Fibrose , Camundongos Transgênicos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Mutação Puntual/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sarcoglicanas/genética , Sarcoglicanas/metabolismo , Sarcolema/metabolismo
7.
JCI Insight ; 4(6)2019 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-30730308

RESUMO

Myotonic dystrophy (DM) is the most common autosomal dominant muscular dystrophy and encompasses both skeletal muscle and cardiac complications. DM is nucleotide repeat expansion disorder in which type 1 (DM1) is due to a trinucleotide repeat expansion on chromosome 19 and type 2 (DM2) arises from a tetranucleotide repeat expansion on chromosome 3. Developing representative models of DM in animals has been challenging due to instability of nucleotide repeat expansions, especially for DM2, which is characterized by nucleotide repeat expansions often greater than 5,000 copies. To investigate mechanisms of human DM, we generated cellular models of DM1 and DM2. We used regulated MyoD expression to reprogram urine-derived cells into myotubes. In this myogenic cell model, we found impaired dystrophin expression, in the presence of muscleblind-like 1 (MBNL1) foci, and aberrant splicing in DM1 but not in DM2 cells. We generated induced pluripotent stem cells (iPSC) from healthy controls and DM1 and DM2 subjects, and we differentiated these into cardiomyocytes. DM1 and DM2 cells displayed an increase in RNA foci concomitant with cellular differentiation. iPSC-derived cardiomyocytes from DM1 but not DM2 had aberrant splicing of known target genes and MBNL sequestration. High-resolution imaging revealed tight association between MBNL clusters and RNA foci in DM1. Ca2+ transients differed between DM1- and DM2 iPSC-derived cardiomyocytes, and each differed from healthy control cells. RNA-sequencing from DM1- and DM2 iPSC-derived cardiomyocytes revealed distinct misregulation of gene expression, as well as differential aberrant splicing patterns. Together, these data support that DM1 and DM2, despite some shared clinical and molecular features, have distinct pathological signatures.


Assuntos
Predisposição Genética para Doença/genética , Proteína MyoD/metabolismo , Distrofia Miotônica/genética , Distrofia Miotônica/patologia , Cálcio/metabolismo , Linhagem Celular , Distrofina/metabolismo , Expressão Gênica , Variação Genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Distrofia Miotônica/classificação , Distrofia Miotônica/urina , Splicing de RNA , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
8.
JCI Insight ; 3(9)2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29720576

RESUMO

Exon skipping uses chemically modified antisense oligonucleotides to modulate RNA splicing. Therapeutically, exon skipping can bypass mutations and restore reading frame disruption by generating internally truncated, functional proteins to rescue the loss of native gene expression. Limb-girdle muscular dystrophy type 2C is caused by autosomal recessive mutations in the SGCG gene, which encodes the dystrophin-associated protein γ-sarcoglycan. The most common SGCG mutations disrupt the transcript reading frame abrogating γ-sarcoglycan protein expression. In order to treat most SGCG gene mutations, it is necessary to skip 4 exons in order to restore the SGCG transcript reading frame, creating an internally truncated protein referred to as Mini-Gamma. Using direct reprogramming of human cells with MyoD, myogenic cells were tested with 2 antisense oligonucleotide chemistries, 2'-O-methyl phosphorothioate oligonucleotides and vivo-phosphorodiamidate morpholino oligomers, to induce exon skipping. Treatment with vivo-phosphorodiamidate morpholino oligomers demonstrated efficient skipping of the targeted exons and corrected the mutant reading frame, resulting in the expression of a functional Mini-Gamma protein. Antisense-induced exon skipping of SGCG occurred in normal cells and those with multiple distinct SGCG mutations, including the most common 521ΔT mutation. These findings demonstrate a multiexon-skipping strategy applicable to the majority of limb-girdle muscular dystrophy 2C patients.


Assuntos
Morfolinos/genética , Sarcoglicanopatias/genética , Sarcoglicanopatias/terapia , Sarcoglicanas/genética , Células Cultivadas , Reprogramação Celular , Éxons , Fibroblastos/metabolismo , Terapia Genética , Humanos , Microscopia de Fluorescência , Mutação , Cultura Primária de Células , Splicing de RNA , Fases de Leitura , Sarcoglicanopatias/metabolismo , Sarcoglicanas/metabolismo , Transdução Genética , Urina/citologia
10.
Skelet Muscle ; 6: 32, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27651888

RESUMO

BACKGROUND: Cellular models of muscle disease are taking on increasing importance with the large number of genes and mutations implicated in causing myopathies and the concomitant need to test personalized therapies. Developing cell models relies on having an easily obtained source of cells, and if the cells are not derived from muscle itself, a robust reprogramming process is needed. Fibroblasts are a human cell source that works well for the generation of induced pluripotent stem cells, which can then be differentiated into cardiomyocyte lineages, and with less efficiency, skeletal muscle-like lineages. Alternatively, direct reprogramming with the transcription factor MyoD has been used to generate myotubes from cultured human fibroblasts. Although useful, fibroblasts require a skin biopsy to obtain and this can limit their access, especially from pediatric populations. RESULTS: We now demonstrate that direct reprogramming of urine-derived cells is a highly efficient and reproducible process that can be used to establish human myogenic cells. We show that this method can be applied to urine cells derived from normal individuals as well as those with muscle diseases. Furthermore, we show that urine-derived cells can be edited using CRISPR/Cas9 technology. CONCLUSIONS: With progress in understanding the molecular etiology of human muscle diseases, having a readily available, noninvasive source of cells from which to generate muscle-like cells is highly useful.


Assuntos
Reprogramação Celular , Desenvolvimento Muscular , Doenças Musculares/fisiopatologia , Proteína MyoD/metabolismo , Urina/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Células Clonais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Edição de Genes , Humanos , Doenças Musculares/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatologia
11.
J Clin Invest ; 126(4): 1236-8, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26999602

RESUMO

Exon skipping uses antisense oligonucleotides (ASOs) to alter transcript splicing for the purpose of rescuing or modulating protein expression. In this issue of the JCI, Lee and colleagues developed and evaluated an ASO-dependent method for treating certain molecularly defined diseases associated with alterations in lamin A/C (LMNA) splicing. Exon skipping by ASOs is gaining traction as a therapeutic strategy, and the use of ASOs is now being applied to bypass mutations and generate modified but functional proteins for an array of genetic disorders.


Assuntos
Processamento Alternativo/efeitos dos fármacos , Lamina Tipo A/biossíntese , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Progéria/tratamento farmacológico , Progéria/metabolismo , RNA Mensageiro/metabolismo , Animais , Humanos
12.
J Clin Invest ; 125(11): 4186-95, 2015 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-26457733

RESUMO

Exon skipping uses antisense oligonucleotides as a treatment for genetic diseases. The antisense oligonucleotides used for exon skipping are designed to bypass premature stop codons in the target RNA and restore reading frame disruption. Exon skipping is currently being tested in humans with dystrophin gene mutations who have Duchenne muscular dystrophy. For Duchenne muscular dystrophy, the rationale for exon skipping derived from observations in patients with naturally occurring dystrophin gene mutations that generated internally deleted but partially functional dystrophin proteins. We have now expanded the potential for exon skipping by testing whether an internal, in-frame truncation of a transmembrane protein γ-sarcoglycan is functional. We generated an internally truncated γ-sarcoglycan protein that we have termed Mini-Gamma by deleting a large portion of the extracellular domain. Mini-Gamma provided functional and pathological benefits to correct the loss of γ-sarcoglycan in a Drosophila model, in heterologous cell expression studies, and in transgenic mice lacking γ-sarcoglycan. We generated a cellular model of human muscle disease and showed that multiple exon skipping could be induced in RNA that encodes a mutant human γ-sarcoglycan. Since Mini-Gamma represents removal of 4 of the 7 coding exons in γ-sarcoglycan, this approach provides a viable strategy to treat the majority of patients with γ-sarcoglycan gene mutations.


Assuntos
Complexo de Proteínas Associadas Distrofina/química , Terapia Genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Oligonucleotídeos Antissenso/uso terapêutico , Engenharia de Proteínas , Sarcoglicanas/genética , Animais , Códon sem Sentido/genética , Diafragma/metabolismo , Diafragma/patologia , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Éxons , Fibrose , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Distrofia Muscular Animal/terapia , Mutação , Miocárdio/metabolismo , Miocárdio/patologia , Oligonucleotídeos Antissenso/farmacologia , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , RNA Mensageiro/química , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/metabolismo , Sarcoglicanas/biossíntese , Sarcoglicanas/química , Sarcoglicanas/deficiência , Sarcolema/metabolismo , Deleção de Sequência
13.
J Neuromuscul Dis ; 1(2): 197-206, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26207203

RESUMO

The New Directions in the Biology and Disease of Skeletal Muscle is a scientific meeting, held every other year, with the stated purpose of bringing together scientists, clinicians, industry representatives and patient advocacy groups to disseminate new discovery useful for treatment inherited forms of neuromuscular disease, primarily the muscular dystrophies. This meeting originated as a response the Muscular Dystrophy Care Act in order to provide a venue for the free exchange of information, with the emphasis on unpublished or newly published data. Highlights of this years' meeting included results from early phase clinical trials for Duchenne Muscular Dystrophy, progress in understanding the epigenetic defects in Fascioscapulohumeral Muscular Dystrophy and new mechanisms of muscle membrane repair. The following is a brief report of the highlights from the conference.

14.
EMBO Mol Med ; 4(7): 633-46, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22517678

RESUMO

Hexokinase-II (HKII) is highly expressed in the heart and can bind to the mitochondrial outer membrane. Since cardiac hypertrophy is associated with a substrate switch from fatty acid to glucose, we hypothesized that a reduction in HKII would decrease cardiac hypertrophy after pressure overload. Contrary to our hypothesis, heterozygous HKII-deficient (HKII(+/-)) mice displayed increased hypertrophy and fibrosis in response to pressure overload. The mechanism behind this phenomenon involves increased levels of reactive oxygen species (ROS), as HKII knockdown increased ROS accumulation, and treatment with the antioxidant N-acetylcysteine (NAC) abrogated the exaggerated response. HKII mitochondrial binding is also important for the hypertrophic effects, as HKII dissociation from the mitochondria resulted in de novo hypertrophy, which was also attenuated by NAC. Further studies showed that the increase in ROS levels in response to HKII knockdown or mitochondrial dissociation is mediated through increased mitochondrial permeability and not by a significant change in antioxidant defenses. Overall, these data suggest that HKII and its mitochondrial binding negatively regulate cardiac hypertrophy by decreasing ROS production via mitochondrial permeability.


Assuntos
Cardiomegalia/metabolismo , Hexoquinase/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Cardiomegalia/patologia , Células Cultivadas , Fibrose , Heterozigoto , Hexoquinase/genética , Hexoquinase/metabolismo , Masculino , Camundongos , Mitocôndrias/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Pressão , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley
15.
J Psychiatry Neurosci ; 36(3): 195-203, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21223646

RESUMO

BACKGROUND: Brain-derived neurotrophic factor (BDNF), tyrosine kinase receptor (trkB-TK+) and glutamic acid decarboxylase (GAD67) mRNA levels have previously been found to be reduced in the prefrontal cortex of patients with schizophrenia. To determine whether this reduction extends to other brain regions, we measured the expression levels of BDNF, trkB-TK+ and GAD67 mRNA in regions of the hippocampus, including the dentate gyrus (DG), cornu ammonis subfields (CA1-4), subiculum and entorhinal cortex (EC) of individuals with schizophrenia, bipolar disorder, major depression and unaffected controls. METHODS: In situ hybridization was performed on postmortem brain tissue obtained from the Stanley Foundation Consortium and analyzed using film-based quantification. RESULTS: Analyses of covariance comparing the expression of mRNA among all groups revealed a significant decrease in BDNF mRNA in CA4 in the bipolar disorder group compared with controls (33%). We found trkB-TK+ mRNA levels to be significantly reduced in CA4 in the schizophrenia group (36%) and in layer II of the EC in the bipolar disorder and major depression groups (28%, 21%, respectively) compared with controls. In addition, GAD67 mRNA levels were reduced in patients with schizophrenia in both the DG (23%) and CA4 (60%) compared with controls. Individuals with major depression also expressed significantly less GAD67 mRNA (44%) compared with controls in CA4 of the hippocampus. LIMITATIONS: It is necessary to account for factors that influence the molecular preservation in postmortem brain tissue, including pH, postmortem interval and tissue storage time. Moreover, there are limitations to the sensitivity of the film-based method of quantification. CONCLUSION: Our findings show abnormal BDNF, trkB-TK+ and GAD67 mRNA expression in the hippocampus of individuals with schizophrenia and mood disorders, indicating that fundamental properties of hippocampal signalling transmission, plasticity and circuitry may be affected in individuals with these major mental illnesses.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Glutamato Descarboxilase/metabolismo , Hipocampo/metabolismo , Transtornos do Humor/metabolismo , Receptor trkB/metabolismo , Esquizofrenia/metabolismo , Adulto , Análise de Variância , Fator Neurotrófico Derivado do Encéfalo/genética , Feminino , Glutamato Descarboxilase/genética , Humanos , Hibridização In Situ , Masculino , Pessoa de Meia-Idade , Transtornos do Humor/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor trkB/genética , Esquizofrenia/genética
16.
Circ Res ; 108(1): 60-9, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21071708

RESUMO

RATIONALE: Cardiomyocytes switch substrate utilization from fatty acid to glucose under ischemic conditions; however, it is unknown how perturbations in glycolytic enzymes affect cardiac response to ischemia/reperfusion (I/R). Hexokinase (HK)II is a HK isoform that is expressed in the heart and can bind to the mitochondrial outer membrane. OBJECTIVE: We sought to define how HKII and its binding to mitochondria play a role in cardiac response and remodeling after I/R. METHODS AND RESULTS: We first showed that HKII levels and its binding to mitochondria are reduced 2 days after I/R. We then subjected the hearts of wild-type and heterozygote HKII knockout (HKII(+/)⁻) mice to I/R by coronary ligation. At baseline, HKII(+/)⁻ mice have normal cardiac function; however, they display lower systolic function after I/R compared to wild-type animals. The mechanism appears to be through an increase in cardiomyocyte death and fibrosis and a reduction in angiogenesis; the latter is through a decrease in hypoxia-inducible factor-dependent pathway signaling in cardiomyocytes. HKII mitochondrial binding is also critical for cardiomyocyte survival, because its displacement in tissue culture with a synthetic peptide increases cell death. Our results also suggest that HKII may be important for the remodeling of the viable cardiac tissue because its modulation in vitro alters cellular energy levels, O2 consumption, and contractility. CONCLUSIONS: These results suggest that reduction in HKII levels causes altered remodeling of the heart in I/R by increasing cell death and fibrosis and reducing angiogenesis and that mitochondrial binding is needed for protection of cardiomyocytes.


Assuntos
Hexoquinase/metabolismo , Proteínas Musculares/metabolismo , Traumatismo por Reperfusão Miocárdica/enzimologia , Miocárdio/enzimologia , Miócitos Cardíacos/enzimologia , Animais , Morte Celular , Metabolismo Energético/genética , Fibrose , Hexoquinase/genética , Camundongos , Mitocôndrias Cardíacas/enzimologia , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/patologia , Proteínas Musculares/genética , Contração Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia , Miócitos Cardíacos/patologia , Consumo de Oxigênio/genética , Fatores de Tempo
17.
PLoS One ; 5(11): e13823, 2010 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21072205

RESUMO

BACKGROUND: Hexokinases (HKs) catalyze the first step in glucose metabolism. Of the three mammalian 100-kDa HK isoforms, HKI and II can bind to mitochondria and protect against cell death. HKIII does not bind mitochondria, and little is known about its regulation or cytoprotective effects. We studied the regulation of HKIII at the transcriptional and protein levels and investigated its role in cellular protection. METHODOLOGY/PRINCIPAL FINDINGS: We show that like HKII, HKIII expression is regulated by hypoxia, but other factors that regulate HKII expression have no effect on HKIII levels. This transcriptional regulation is partially dependent on hypoxia-inducible factor (HIF) signaling. We also demonstrate regulation at the protein level, as mutations in putative N-terminal substrate binding residues altered C-terminal catalytic activity, suggesting that HKIII activity is governed, in part, by interactions between these two domains. Overexpression of HKIII reduced oxidant-induced cell death, increased ATP levels, decreased the production of reactive oxygen species (ROS), and preserved mitochondrial membrane potential. HKIII overexpression was also associated with higher levels of transcription factors that regulate mitochondrial biogenesis, and greater total mitochondrial DNA content. Attempts to target HKIII to the mitochondria by replacing its N-terminal 32-amino-acid sequence with the mitochondrial-targeting sequence of HKII led to protein aggregation, suggesting that this region is necessary to maintain proper protein folding and solubility. CONCLUSIONS/SIGNIFICANCE: These results suggest that HKIII is regulated by hypoxia and there are functional interactions between its two halves. Furthermore, HKIII exerts protective effects against oxidative stress, perhaps by increasing ATP levels, reducing oxidant-induced ROS production, preserving mitochondrial membrane potential, and increasing mitochondrial biogenesis.


Assuntos
Citoproteção/genética , Regulação Enzimológica da Expressão Gênica , Hexoquinase/genética , Trifosfato de Adenosina/metabolismo , Animais , Western Blotting , Domínio Catalítico/genética , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Hexoquinase/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Potencial da Membrana Mitocondrial/fisiologia , Microscopia Confocal , Mutação , Oxidantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Transfecção
18.
J Psychiatr Res ; 43(11): 970-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19321177

RESUMO

Reduced levels of glutamic acid decarboxylase(67) (GAD(67)), an essential enzyme for GABA synthesis, is one of the most consistent gene expression changes found in the frontal cortex of patients with schizophrenia. Recently this reduction has been shown to extend to other areas including primary sensory, primary motor and anterior cingulate (ACC) cortices. To determine the extent to which additional cortical and subcortical regions may be affected in schizophrenia, we measured the level of GAD(67) mRNA in previously unexplored areas including the orbitofrontal (OFC) and superior temporal (STG) cortices as well as the caudate, putamen, nucleus accumbens, medial dorsal thalamus and anterior thalamus using in situ hybridization. We also examined GAD(67) mRNA levels in all these regions in individuals with bipolar disorder and major depression. ANCOVA comparing GAD(67) mRNA levels in all four diagnostic groups revealed a significant reduction (approximately 30%) in layers III and IV of the OFC of patients with schizophrenia and bipolar disorder. A priori t-tests comparing GAD(67) mRNA levels between the schizophrenia and control groups revealed significant reductions in the ACC, STG, striatum and thalamus. These findings suggest that there may be a widespread reduction in GABA neurotransmission due to a decrease in the synthesis of GAD(67) in subjects with psychiatric disorders. The resulting decrease in inhibitory tone across multiple brain areas may contribute to the psychotic behavior observed in patients with schizophrenia and bipolar disorder.


Assuntos
Encéfalo/enzimologia , Encéfalo/patologia , Regulação Enzimológica da Expressão Gênica , Glutamato Descarboxilase/metabolismo , Transtornos do Humor/enzimologia , RNA Mensageiro/metabolismo , Esquizofrenia/enzimologia , Adulto , Idoso , Análise de Variância , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos do Humor/patologia , Esquizofrenia/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA